151
|
Wang T, Li Y, Wang W, Tuerhanjiang A, Wu Z, Yang R, Yuan M, Ma D, Wang W, Wang S. Twist2, the key Twist isoform related to prognosis, promotes invasion of cervical cancer by inducing epithelial-mesenchymal transition and blocking senescence. Hum Pathol 2014; 45:1839-46. [PMID: 24974259 DOI: 10.1016/j.humpath.2014.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/04/2014] [Accepted: 05/07/2014] [Indexed: 01/28/2023]
Abstract
In response to tumor development, cells initially undergo invasion and metastasis followed by epithelial-mesenchymal transition (EMT, a process by which cells acquire motility) and overriding senescence (an endogenous defense mechanism against tumor progression). Oncogenic activation of Twist1 and Twist2 is essential for EMT and senescence; however, little is known about the specific contributions of Twist1 versus Twist2 to prognosis, metastasis, and the mechanism underlying cervical carcinoma. Here, we investigated the similarities and differences between Twist1 and Twist2 in assessing prognosis and promoting invasion and metastasis of cervical carcinoma as well as their roles in the underlying molecular mechanisms. By monitoring the survival of 144 clinical cervical cancer patients, we demonstrated that Twist2 shows more effective predictive performance compared with Twist1 and is more closely correlated with International Federation of Gynecology and Obstetrics stage and lymph node metastasis. Compared with Twist1, Twist2 more strongly promotes invasivity and motility by inducing EMT and overriding senescence. Differences between Twist1 and Twist2 in regulating senescence and the cell cycle might be due to their individual roles in regulating the cyclin D1/cyclin dependent kinase 4 (Cdk4) pathway. Overall, our data indicate that Twist2 is the key Twist isoform coupling aberrant signals from EMT to senescence and is an important candidate biomarker for cervical cancer prognosis.
Collapse
Affiliation(s)
- Tian Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Abidan Tuerhanjiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Zhangying Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Runfeng Yang
- Department of Gynecologic Oncology, Tumor Hospital of Hubei Provincial, Wuhan, Hubei, 430079, PR China
| | - Ming Yuan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Ding Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China.
| | - Wei Wang
- Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China.
| |
Collapse
|
152
|
Warner DR, Mukhopadhyay P, Brock G, Webb CL, Michele Pisano M, Greene RM. MicroRNA expression profiling of the developing murine upper lip. Dev Growth Differ 2014; 56:434-47. [PMID: 24849136 DOI: 10.1111/dgd.12140] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 03/28/2014] [Accepted: 04/01/2014] [Indexed: 12/21/2022]
Abstract
Clefts of the lip and palate are thought to be caused by genetic and environmental insults but the role of epigenetic mechanisms underlying this common birth defect are unknown. We analyzed the expression of over 600 microRNAs in the murine medial nasal and maxillary processes isolated on GD10.0-GD11.5 to identify those expressed during development of the upper lip and analyzed spatial expression of a subset. A total of 142 microRNAs were differentially expressed across gestation days 10.0-11.5 in the medial nasal processes, and 66 in the maxillary processes of the first branchial arch with 45 common to both. Of the microRNAs exhibiting the largest percent increase in both facial processes were five members of the Let-7 family. Among those with the greatest decrease in expression from GD10.0 to GD11.5 were members of the microRNA-302/367 family that have been implicated in cellular reprogramming. The distribution of expression of microRNA-199a-3p and Let-7i was determined by in situ hybridization and revealed widespread expression in both medial nasal and maxillary facial process, while that for microRNA-203 was much more limited. MicroRNAs are dynamically expressed in the tissues that form the upper lip and several were identified that target mRNAs known to be important for its development, including those that regulate the two main isoforms of p63 (microRNA-203 and microRNA-302/367 family). Integration of these data with corresponding proteomic datasets will lead to a greater appreciation of epigenetic regulation of lip development and provide a better understanding of potential causes of cleft lip.
Collapse
Affiliation(s)
- Dennis R Warner
- Department of Molecular, Cellular, and Craniofacial Biology, University of Louisville Birth Defects Center, Louisville, Kentucky, USA
| | | | | | | | | | | |
Collapse
|
153
|
Luo Z, Wen G, Wang G, Pu X, Ye S, Xu Q, Wang W, Xiao Q. MicroRNA-200C and -150 play an important role in endothelial cell differentiation and vasculogenesis by targeting transcription repressor ZEB1. Stem Cells 2014; 31:1749-62. [PMID: 23765923 DOI: 10.1002/stem.1448] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 04/30/2013] [Accepted: 05/06/2013] [Indexed: 12/19/2022]
Abstract
To investigate the role of miRNA in controlling human embryonic stem (hES) cell differentiation toward the endothelial lineage and chick embryonic blood vessel formation, undifferentiated hES cells were first cultured on Matrigel-coated flasks and in endothelial cell growth medium-2 (EGM-2) to initiate endothelial cell (EC) differentiation. CD146(+) cells were isolated from differentiating hES cells and expanded in vitro. The in vitro expanded CD146(+) cells were positive for EC markers, capable of Ac-LDL uptake, lectin binding, and the formation of vascular structures in vitro and in vivo. miRNA gain/loss-of-function analyses revealed that miR-150 and miR-200c were crucial in EC differentiation. Transcriptional repressor zinc finger E-box-binding homeobox 1 (ZEB1) was identified as the communal target gene of miRNA-200C and -150, and inhibition of ZEB1 was required for miRNA-200C or -150 mediated EC gene expressions. Moreover, we demonstrated that ZEB1 could transcriptionally repress EC gene expression through direct binding to promoters of EC genes. Finally, we also demonstrated that miRNA-200c and -150 played an important role in chick embryonic blood vessel formation by in vivo inhibition of miRNA-200C or -150 in developing chick embryos, and blocking ZEB1 signaling in CD146-positive cells could rescue the inhibitory effects of miR-200c inhibiton in in vivo vasculogenesis. Our findings revealed that miR-150 and miR-200c play an important role in human endothelial lineage specification and chick embryonic vasculogenesis by targeting ZEB1.
Collapse
Affiliation(s)
- Zhenling Luo
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom; Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
154
|
Rb1 family mutation is sufficient for sarcoma initiation. Nat Commun 2014; 4:2650. [PMID: 24150016 DOI: 10.1038/ncomms3650] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/19/2013] [Indexed: 12/16/2022] Open
Abstract
It is thought that genomic instability precipitated by Rb1 pathway loss rapidly triggers additional cancer gene mutations, accounting for rapid tumour onset following Rb1 mutation. However, recent whole-genome sequencing of retinoblastomas demonstrated little genomic instability, but instead suggested rapid epigenetic activation of cancer genes. These results raise the possibility that loss of the Rb1 pathway, which is a hallmark of cancers, might be sufficient for cancer initiation. Yet, mutation of the Rb1 family or inactivation of the Rb1 pathway in primary cells has proven insufficient for tumour initiation. Here we demonstrate that traditional nude mouse assays impose an artificial anoikis and proliferation barrier that prevents Rb1 family mutant fibroblasts from initiating tumours. By circumventing this barrier, we show that primary fibroblasts with only an Rb1 family mutation efficiently form sarcomas in nude mice, and a Ras-ZEB1-Akt pathway then causes transition of these tumours to an invasive phenotype.
Collapse
|
155
|
Zerbini LF, Bhasin MK, de Vasconcellos JF, Paccez JD, Gu X, Kung AL, Libermann TA. Computational repositioning and preclinical validation of pentamidine for renal cell cancer. Mol Cancer Ther 2014; 13:1929-1941. [PMID: 24785412 DOI: 10.1158/1535-7163.mct-13-0750] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although early stages of clear cell renal cell carcinoma (ccRCC) are curable, survival outcome for metastatic ccRCC remains poor. We previously established a highly accurate signature of differentially expressed genes that distinguish ccRCC from normal kidney. The purpose of this study was to apply a new individualized bioinformatics analysis (IBA) strategy to these transcriptome data in conjunction with Gene Set Enrichment Analysis of the Connectivity Map (C-MAP) database to identify and reposition FDA-approved drugs for anticancer therapy. Here, we demonstrate that one of the drugs predicted to revert the RCC gene signature toward normal kidney, pentamidine, is effective against RCC cells in culture and in a RCC xenograft model. ccRCC-specific gene expression signatures of individual patients were used to query the C-MAP software. Eight drugs with negative correlation and P-value <0.05 were analyzed for efficacy against RCC in vitro and in vivo. Our data demonstrate consistency across most patients with ccRCC for the set of high-scoring drugs. Most of the selected high-scoring drugs potently induce apoptosis in RCC cells. Several drugs also demonstrate selectivity for Von Hippel-Lindau negative RCC cells. Most importantly, at least one of these drugs, pentamidine, slows tumor growth in the 786-O human ccRCC xenograft mouse model. Our findings suggest that pentamidine might be a new therapeutic agent to be combined with current standard-of-care regimens for patients with metastatic ccRCC and support our notion that IBA combined with C-MAP analysis enables repurposing of FDA-approved drugs for potential anti-RCC therapy.
Collapse
Affiliation(s)
- Luiz Fernando Zerbini
- International Center for Genetic Engineering and Biotechnology (ICGEB), Cancer Genomics Group and Division of Medical Biochemistry, University of Cape Town, Cape Town, South Africa.,BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Manoj K Bhasin
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Jaira F de Vasconcellos
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Juliano D Paccez
- International Center for Genetic Engineering and Biotechnology (ICGEB), Cancer Genomics Group and Division of Medical Biochemistry, University of Cape Town, Cape Town, South Africa
| | - Xuesong Gu
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Andrew L Kung
- Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Towia A Libermann
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| |
Collapse
|
156
|
Jang MS, Roldan AN, Frausto RF, Aldave AJ. Posterior polymorphous corneal dystrophy 3 is associated with agenesis and hypoplasia of the corpus callosum. Vision Res 2014; 100:88-92. [PMID: 24780443 DOI: 10.1016/j.visres.2014.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 03/14/2014] [Accepted: 04/02/2014] [Indexed: 11/16/2022]
Abstract
Posterior polymorphous corneal dystrophy (PPCD) is a dominantly inherited disorder of the corneal endothelium that has been associated with mutations in the zinc-finger E-box binding homeobox 1 gene (ZEB1) gene in approximately one-third of affected families. While the corneal dystrophies have traditionally been considered isolated disorders of the corneal endothelium, we have recently identified two cases of maldevelopment of the corpus callosum in unrelated individuals with PPCD. The proband of the first family was diagnosed shortly after birth with agenesis of the corpus callosum and several other developmental abnormalities. Karyotype, FISH and whole genome copy number variant analyses were normal. She was subsequently diagnosed with PPCD, prompting screening of the ZEB1 gene, which identified a novel deletion (c.449delG; p.(Gly150Alafs*36)) present in the heterozygous state that was not identified in either unaffected parent. The proband of the second family was diagnosed several months after birth with thinning of the corpus callosum and PPCD. Whole genome copy number variant analysis revealed a 1.79 Mb duplication of 17q12 in the proband and her father and brother, neither of whom had PPCD. ZEB1 sequencing identified a novel deletion (c.1913-1914delCA; p.(Ser638Cysfs*5)) present in the heterozygous state, which was also identified in the proband's affected mother. Thus, we report the first two cases of the association of PPCD with a developmental abnormality of the brain, in this case maldevelopment of the corpus callosum.
Collapse
Affiliation(s)
- Michelle S Jang
- The Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Ashley N Roldan
- The Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Ricardo F Frausto
- The Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Anthony J Aldave
- The Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
157
|
Xie G, Ji A, Yuan Q, Jin Z, Yuan Y, Ren C, Guo Z, Yao Q, Yang K, Lin X, Chen L. Tumour-initiating capacity is independent of epithelial-mesenchymal transition status in breast cancer cell lines. Br J Cancer 2014; 110:2514-23. [PMID: 24755887 PMCID: PMC4021510 DOI: 10.1038/bjc.2014.153] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 12/18/2022] Open
Abstract
Background: Epithelial–mesenchymal transition (EMT) and cancer stem cells (CSCs) are considered to be crucial for cancer biology. The purpose of this study was to determine whether EMT directly led to the acquisition of tumour-initiating capacity in breast cancer cell lines. Methods: Epithelial–mesenchymal transition was induced in five breast cancer cell lines and one normal breast cell line by EMT-related cytokine stimulation. Mesenchymal–epithelial transition (MET) was induced by stably overexpressing miR-200c in three mesenchymal-like breast cancer cell lines. Molecular expression and cell function analysis were performed to evaluate the effect of EMT or MET on tumour-initiating capacity and other biological characteristics. Results: The induction of EMT did not enhance tumour-initiating capacity but, instead, conferred a CD44+/CD24−/low phenotype as well as cell proliferation, migration, and resistance to doxorubicin and radiation on breast cancer cell lines. Furthermore, MET did not lead to inhibition or loss of the tumour-initiating capacity in mesenchymal-like breast cancer cell lines, but it markedly attenuated other malignant properties, including proliferation, invasion, and resistance to therapy. Conclusions: Epithelial–mesenchymal transition does not alter tumour-initiating capacity of breast cancer cells but some other biological characteristics. Therefore, EMT and tumour-initiating capacity may not be directly linked in breast cancer cell lines.
Collapse
Affiliation(s)
- G Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - A Ji
- Department of Pharmaceutical Science, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, PR China
| | - Q Yuan
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Z Jin
- Department of Pharmacy, Jining First People's Hospital, Jining, Shandong 272111, PR China
| | - Y Yuan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - C Ren
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Z Guo
- Breast Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Q Yao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - K Yang
- Department of Neurosurgery, Institute of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - X Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - L Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
158
|
Jain K, Basu A. Protein Kinase C-ε Promotes EMT in Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2014; 8:61-7. [PMID: 24701121 PMCID: PMC3972078 DOI: 10.4137/bcbcr.s13640] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/15/2013] [Accepted: 12/16/2013] [Indexed: 01/17/2023]
Abstract
Protein kinase C (PKC), a family of serine/threonine kinases, plays critical roles in signal transduction and cell regulation. PKCε, a member of the novel PKC family, is known to be a transforming oncogene and a tumor biomarker for aggressive breast cancers. In this study, we examined the involvement of PKCε in epithelial to mesenchymal transition (EMT), the process that leads the way to metastasis. Overexpression of PKCε was sufficient to induce a mesenchymal phenotype in non-tumorigenic mammary epithelial MCF-10 A cells. This was accompanied by a decrease in the epithelial markers, such as E-cadherin, zonula occludens (ZO)-1, and claudin-1, and an increase in mesenchymal marker vimentin. Transforming growth factor β (TGFβ) induced Snail expression and mesenchymal morphology in MCF-10 A cells, and these effects were partially reversed by the PKCε knockdown. PKCε also mediated cell migration and anoikis resistance, which are hallmarks of EMT. Thus, our study demonstrates that PKCε is an important mediator of EMT in breast cancer.
Collapse
Affiliation(s)
- Kirti Jain
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Institute for Cancer Research, Fort Worth, TX, USA. ; Focused on Resources for her Health Education and Research, Fort Worth, TX, USA
| | - Alakananda Basu
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, Institute for Cancer Research, Fort Worth, TX, USA. ; Focused on Resources for her Health Education and Research, Fort Worth, TX, USA
| |
Collapse
|
159
|
Bronsert P, Kohler I, Timme S, Kiefer S, Werner M, Schilling O, Vashist Y, Makowiec F, Brabletz T, Hopt UT, Bausch D, Kulemann B, Keck T, Wellner UF. Prognostic significance of Zinc finger E-box binding homeobox 1 (ZEB1) expression in cancer cells and cancer-associated fibroblasts in pancreatic head cancer. Surgery 2014; 156:97-108. [PMID: 24929761 DOI: 10.1016/j.surg.2014.02.018] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 02/24/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterized by an aggressive biology and poor prognosis. Experimental evidence has suggested a role for the transcriptional repressor Zinc finger E-box binding homeobox 1 (ZEB1) in epithelial-mesenchymal transition, invasion, and metastasis in PDAC. ZEB1 expression has been observed in cancer cells as well as stromal fibroblasts. Our study aimed to evaluate the prognostic value of ZEB1 expression in PDAC tissue. METHODS Patient baseline and follow-up data were extracted from a prospectively maintained database. After clinicopathologic re-review, serial sliced tissue slides were immunostained for ZEB1, E-cadherin, vimentin, and pan-cytokeratin. ZEB1 expression in cancer cells and adjacent stromal fibroblasts was graded separately and correlated to routine histopathologic parameters and survival after resection. RESULTS A total of 117 cases of PDAC were included in the study. High ZEB1 expression in cancer cells and in stromal cancer-associated fibroblasts was associated with poor prognosis. There was also a trend for poor prognosis with a lymph node ratio of greater than 0.10. In line with its role as an inducer of epithelial-mesenchymal transition, ZEB1 expression in cancer cells was positively correlated with Vimentin expression and negatively with E-Cadherin expression. In multivariate analysis, stromal ZEB1 expression grade was the only independent factor of survival after resection. CONCLUSION Our data suggest that ZEB1 expression in cancer cells as well as in stromal fibroblasts are strong prognostic factors in PDAC. Stromal ZEB1 expression is identified for the first time as an independent predictor of survival after resection of PDAC. This observation suggests that therapies targeting ZEB1 and its downstream pathways could hit both cancer cells and supporting cancer-associated fibroblasts.
Collapse
Affiliation(s)
- Peter Bronsert
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany; Comprehensive Cancer Center, Freiburg, Germany.
| | - Ilona Kohler
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Sylvia Timme
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Selina Kiefer
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Martin Werner
- Institute of Pathology, University Medical Center Freiburg, Freiburg, Germany; Comprehensive Cancer Center, Freiburg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Schilling
- Institute for Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Yogesh Vashist
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | - Frank Makowiec
- Comprehensive Cancer Center, Freiburg, Germany; Clinic for General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany
| | - Thomas Brabletz
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany; Clinic for General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany
| | - Ulrich T Hopt
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany
| | - Dirk Bausch
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany; Clinic for Surgery, University Clinic Schleswig-Holstein Campus, Lübeck, Germany
| | - Birte Kulemann
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany
| | - Tobias Keck
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany; Clinic for Surgery, University Clinic Schleswig-Holstein Campus, Lübeck, Germany
| | - Ulrich F Wellner
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany; Clinic for Surgery, University Clinic Schleswig-Holstein Campus, Lübeck, Germany
| |
Collapse
|
160
|
Wen FC, Chang TW, Tseng YL, Lee JC, Chang MC. hRAD9 functions as a tumor suppressor by inducing p21-dependent senescence and suppressing epithelial-mesenchymal transition through inhibition of Slug transcription. Carcinogenesis 2014; 35:1481-90. [PMID: 24403312 DOI: 10.1093/carcin/bgu009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Senescence and epithelial-mesenchymal transition (EMT) have opposing roles in tumor progression, in that, one is a barrier against tumorigenesis, whereas the other is required for invasive malignancies. Here, we report that the DNA damage response (DDR) protein hRAD9 contributes to induction of senescence and inhibition of EMT. Our data show that hRAD9 is frequently downregulated in breast and lung cancers. Loss of hRAD9 expression is associated with tumor stage in breast and lung cancers, as well as with acquisition of an invasive phenotype. Ectopic hRAD9 expression in highly invasive cancer cell lines, H1299 and MDA-MB 231, with low endogenous hRAD9 induced senescence by upregulation of nuclear p21, independent of the p53 status. Ectopic expression of hRAD9 also significantly attenuated cellular migration and invasion in vitro and tumor growth in a xenograft mouse model in vivo. In contrast, silencing hRAD9 in lower invasive cancer cell lines, A549 and MCF7, with high endogenous hRAD9 dramatically increased their migration and invasion abilities, and simultaneously activated EMT. Knockdown of hRAD9 increased, whereas ectopic expression of hRAD9 decreased, the expression of Slug. Moreover, hRAD9 directly bound to the promoter region of slug gene and repressed its transcriptional activity. Taken together, these results suggest that hRAD9 is a potential tumor suppressor in breast and lung cancers and that it is likely to function by upregulating p21 and inhibiting Slug to regulate tumorigenesis.
Collapse
Affiliation(s)
- Fan-Chih Wen
- Institute of Basic Medical Sciences and Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tsai-Wang Chang
- Department of Surgery, National Cheng Kung University Medical College and Hospital, Tainan 70101, Taiwan and
| | - Yau-Lin Tseng
- Department of Surgery, National Cheng Kung University Medical College and Hospital, Tainan 70101, Taiwan and
| | - Janq-Chang Lee
- Department of Surgery, National Cheng Kung University Medical College and Hospital, Tainan 70101, Taiwan and
| | - Ming-Chung Chang
- Institute of Basic Medical Sciences and Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan, Department of Nutrition, College of Medicine and Nursing, Hung Kuang University, Taichung 43302, Taiwan
| |
Collapse
|
161
|
Liu Y, Sánchez-Tilló E, Lu X, Huang L, Clem B, Telang S, Jenson AB, Cuatrecasas M, Chesney J, Postigo A, Dean DC. The ZEB1 transcription factor acts in a negative feedback loop with miR200 downstream of Ras and Rb1 to regulate Bmi1 expression. J Biol Chem 2013; 289:4116-25. [PMID: 24371144 DOI: 10.1074/jbc.m113.533505] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ras mutations are frequent in cancer cells where they drive proliferation and resistance to apoptosis. However in primary cells, mutant Ras instead can cause oncogene-induced senescence, a tumor suppressor function linked to repression of the polycomb factor Bmi1, which normally regulates cell cycle inhibitory cyclin-dependent kinase inhibitors (cdki). It is unclear how Ras causes repression of Bmi1 in primary cells to suppress tumor formation while inducing the gene in cancer cells to drive tumor progression. Ras also induces the EMT transcription factor ZEB1 to trigger tumor invasion and metastasis. Beyond its well-documented role in EMT, ZEB1 is important for maintaining repression of cdki. Indeed, heterozygous mutation of ZEB1 is sufficient for elevated cdki expression, leading to premature senescence of primary cells. A similar phenotype is evident with Bmi1 mutation. We show that activation of Rb1 in response to mutant Ras causes dominant repression of ZEB1 in primary cells, but loss of the Rb1 pathway is a hallmark of cancer cells and in the absence of such Rb1 repression Ras induces ZEB1 in cancer cells. ZEB1 represses miR-200 in the context of a mutual repression loop. Because miR-200 represses Bmi1, induction of ZEB1 leads to induction of Bmi1. Rb1 pathway status then dictates the opposing effects of mutant Ras on the ZEB1-miR-200 loop in primary versus cancer cells. This loop not only triggers EMT, surprisingly we show it acts downstream of Ras to regulate Bmi1 expression and thus the critical decision between oncogene-induced senescence and tumor initiation.
Collapse
Affiliation(s)
- Yongqing Liu
- From the Molecular Targets Program, James Brown Cancer Center
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Mohr AM, Bailey JM, Lewallen ME, Liu X, Radhakrishnan P, Yu F, Tapprich W, Hollingsworth MA. MUC1 regulates expression of multiple microRNAs involved in pancreatic tumor progression, including the miR-200c/141 cluster. PLoS One 2013; 8:e73306. [PMID: 24143167 PMCID: PMC3797065 DOI: 10.1371/journal.pone.0073306] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 07/18/2013] [Indexed: 01/08/2023] Open
Abstract
MUC1 is a transmembrane glycoprotein that modulates transcription via its cytoplasmic domain. We evaluated the capacity of MUC1 to regulate the global transcription of microRNAs in pancreatic cancer cells expressing MUC1. Results indicated that MUC1 regulated expression of at least 103 microRNAs. We evaluated further regulation of the microRNA transcript cluster miR-200c/141, which was among the most highly regulated microRNAs. We found that MUC1 directly interacted with ZEB1, a known transcriptional repressor of the miR-200c/141 cluster, at the promoter of miR-200c/141, and further reduced transcript production. These data indicate that signaling through MUC1 influences cancer progression by regulating transcription of microRNAs that are associated with the process of metastasis.
Collapse
Affiliation(s)
- Ashley M. Mohr
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jennifer M. Bailey
- Department of Surgery, the Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Michelle E. Lewallen
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Xiang Liu
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Fang Yu
- College of Public Health Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - William Tapprich
- Department of Biology, University of Nebraska at Omaha, Omaha, Nebraska, United States of America
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
163
|
Sánchez-Tilló E, Fanlo L, Siles L, Montes-Moreno S, Moros A, Chiva-Blanch G, Estruch R, Martinez A, Colomer D, Győrffy B, Roué G, Postigo A. The EMT activator ZEB1 promotes tumor growth and determines differential response to chemotherapy in mantle cell lymphoma. Cell Death Differ 2013; 21:247-57. [PMID: 24013721 DOI: 10.1038/cdd.2013.123] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 07/19/2013] [Accepted: 08/02/2013] [Indexed: 12/28/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a B-cell malignancy characterized by a poor response to treatment and prognosis. Constitutive activation of different signaling pathways in subsets of MCLs, through genetic and/or nongenetic alterations, endows tumor cells with enhanced proliferation and reduced apoptosis. The canonical Wnt pathway (β-catenin/TCF-LEF), implicated in the pathogenesis of numerous cancers, is constitutively active in half of MCLs. Here, we show that ZEB1, a transcription factor better known for promoting metastasis in carcinomas, is expressed in primary MCLs with active Wnt signaling. ZEB1 expression in MCL cells depends on Wnt, being downregulated by β-catenin knockdown or blocking of Wnt signaling by salinomycin. Knockdown of ZEB1 reduces in vitro cell viability and proliferation in MCL cells, and, importantly, tumor growth in mouse xenograft models. ZEB1 activates proliferation-associated (HMGB2, UHRF1, CENPF, MYC, MKI67, and CCND1) and anti-apoptotic (MCL1, BCL2, and BIRC5) genes and inhibits pro-apoptotic ones (TP53, BBC3, PMAIP1, and BAX). We show that ZEB1 expression in MCL cells determines differential resistance to chemotherapy drugs and regulates transporters involved in drug influx/efflux. Downregulation of ZEB1 by salinomycin increases the sensitivity of MCL cells to the cytotoxic effect of doxorubicin, cytarabine and gemcitabine. Lastly, salinomycin and doxorubicin display a synergistic effect in established and primary MCL cells. These results identify ZEB1 in MCL where it promotes cell proliferation, enhanced tumor growth and a differential response to chemotherapy drugs. ZEB1 could thus potentially become a predictive biomarker and therapeutic target in this lymphoma.
Collapse
Affiliation(s)
- E Sánchez-Tilló
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, CIBERehd, Barcelona 08036, Spain
| | - L Fanlo
- 1] Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, CIBERehd, Barcelona 08036, Spain [2] Master Program in Biomedical Research, University Pompeu Fabra, Barcelona 08002, Spain
| | - L Siles
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, CIBERehd, Barcelona 08036, Spain
| | - S Montes-Moreno
- Department of Pathology and Group of Cancer Genomics, Hospital Marques de Valdecilla, IFIMAV, Santander 39008, Spain
| | - A Moros
- Hematopathology Unit, Hospital Clinic, IDIBAPS, Barcelona 08036, Spain
| | - G Chiva-Blanch
- 1] Department of Internal Medicine, Hospital Clinic, Barcelona 08036, Spain [2] CIBERobn, ISCIII, Santiago de Compostela 15706, Spain
| | - R Estruch
- 1] Department of Internal Medicine, Hospital Clinic, Barcelona 08036, Spain [2] CIBERobn, ISCIII, Santiago de Compostela 15706, Spain
| | - A Martinez
- Department of Pathology, Hospital Clinic, Barcelona 08036, Spain
| | - D Colomer
- Hematopathology Unit, Hospital Clinic, IDIBAPS, Barcelona 08036, Spain
| | - B Győrffy
- Research Lab for Pediatrics and Nephrology, Hungarian Academy of Sciences, Semmelweis University, 1st Department of Pediatrics, Budapest 1083, Hungary
| | - G Roué
- Hematopathology Unit, Hospital Clinic, IDIBAPS, Barcelona 08036, Spain
| | - A Postigo
- 1] Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, CIBERehd, Barcelona 08036, Spain [2] James Graham Brown Cancer Center, Louisville, KY 40202, USA [3] ICREA, Barcelona 08010, Spain
| |
Collapse
|
164
|
Jackstadt R, Jung P, Hermeking H. AP4 directly downregulates p16 and p21 to suppress senescence and mediate transformation. Cell Death Dis 2013; 4:e775. [PMID: 23949224 PMCID: PMC3763444 DOI: 10.1038/cddis.2013.282] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/02/2013] [Accepted: 07/04/2013] [Indexed: 12/14/2022]
Abstract
Here we analyzed the function of the c-MYC-inducible basic helix–loop–helix leucine-zipper transcription factor AP4 in AP4-deficient mouse embryo fibroblasts (MEFs). Loss of AP4 resulted in premature senescence and resistance towards immortalization. Senescence was accompanied by induction of the cyclin-dependent kinase inhibitor-encoding genes p16, a known tumor suppressor, and p21, a previously described target for repression by AP4. Notably, AP4 directly repressed p16 expression via conserved E-box motifs in MEFs and human diploid fibroblasts. Senescence caused by AP4-deficiency was prevented by depletion of p16 and/or p21, demonstrating that these factors mediate senescence caused by AP4 loss. As senescence induced by the loss of AP4 was rescued by ectopic AP4, secondary lesions were not involved in causing premature senescence. Activation of c-MYC resulted in repression of p21 and p16 in AP4+/+, but not in AP4−/− MEFs. Furthermore, after combined expression of c-MYC and mutant RAS in MEFs, AP4 was required for colony formation, anchorage-independent growth and tumor formation in mice. In addition, combined ectopic expression of AP4 and mutant RAS in MEFs resulted in colony formation. However, additional loss of the p53 tumor suppressor was necessary for anchorage-independent growth and tumor formation of MEFs by combined AP4 and mutant RAS expression. In conclusion, this study identified AP4 as an oncogenic antagonist of cellular senescence. AP4 achieves this effect by direct repression of p16 and p21, and may thereby critically contribute to c-MYC function and tumor progression.
Collapse
Affiliation(s)
- R Jackstadt
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-Universität München, Thalkirchner Strasse 36, D-80337 Munich, Germany
| | | | | |
Collapse
|
165
|
Abstract
The aim of this review was to provide an evidenced-based review of the genetic basis of the corneal endothelial dystrophies. A review of the English language peer-reviewed literature describing the molecular genetic basis of posterior polymorphous corneal dystrophy (PPCD), congenital hereditary endothelial dystrophy (CHED), Fuchs endothelial corneal dystrophy (FECD) and X-linked endothelial corneal dystrophy (XECD) was performed. Mutations in several genes have been implicated as playing a pathogenic role in the corneal endothelial dystrophies: VSX1 mutations in PPCD1; COL8A2 mutations in PPCD2 and FECD; ZEB1 mutations in PPCD3 and FECD; and SLC4A11 mutations in CHED2 and FECD. However, linkage, association and familial segregation analyses support a role of only one gene in each corneal endothelial dystrophy: ZEB1 in PPCD3, SLC4A11 in CHED2 and COL8A2 in FECD (early onset). In addition, insufficient evidence exists to consider the autosomal dominant form of CHED (CHED1) as distinct from PPCD. An accurate classification of the corneal endothelial dystrophies requires a critical review of the evidence to support the role of each suggested chromosomal locus, gene and genetic mutation associated with a corneal endothelial dystrophy. Only after the separation of evidence from opinion is performed can a critical examination of the molecular pathways that lead to endothelial dysfunction in each of these disorders be accurately performed.
Collapse
Key Words
- corneal dystrophy, Fuchs endothelial, 1
- corneal dystrophy, Fuchs endothelial, 2
- corneal dystrophy, Fuchs endothelial, early onset
- corneal dystrophy, Fuchs endothelial, late onset
- corneal dystrophy, posterior polymorphous, 1
- corneal dystrophy, posterior polymorphous, 2
- corneal dystrophy, posterior polymorphous, 3
- corneal endothelial dystrophy 1
- corneal endothelial dystrophy 2
Collapse
Affiliation(s)
- A J Aldave
- The Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095–7003, USA.
| | | | | |
Collapse
|
166
|
Liu Y, Mukhopadhyay P, Pisano MM, Lu X, Huang L, Lu Q, Dean DC. Repression of Zeb1 and hypoxia cause sequential mesenchymal-to-epithelial transition and induction of aid, Oct4, and Dnmt1, leading to immortalization and multipotential reprogramming of fibroblasts in spheres. Stem Cells 2013; 31:1350-62. [PMID: 23554223 PMCID: PMC4265806 DOI: 10.1002/stem.1382] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/06/2013] [Indexed: 12/26/2022]
Abstract
In this study, we demonstrate that sphere formation triggers immortalization and stable reprogramming of mouse fibroblasts. Cell contact signaling in spheres causes downregulation of the epithelial-to-mesenchymal transition transcription factor Zeb1 leading to rapid mesenchymal-to-epithelial transition. Hypoxia within spheres together with loss of Zeb1 repression synergize to cause superinduction of Hif1a, which in turn leads to induction of the DNA demethylase Aid/Aicda, demethylation of the Oct4 promoter/enhancer and multipotency. Oct4 and Nanog expression diminish when cells are removed from the hypoxic environment of spheres and placed in monolayer culture, but the cells retain multipotential capacity, demonstrating stable reprogramming and a gene expression pattern resembling adult stem cells. Oct4 has been shown to induce Dnmt1 in mesenchymal stem cells, and we link Oct4 and Dnmt1 to silencing of cell cycle inhibitory cyclin dependent kinase inhibitors and Arf, and immortalization of the reprogrammed fibroblasts. Sphere formation then represents a novel and rapid protocol for immortalization and stable reprogramming of fibroblasts to multipotency that does not require exogenous expression of a stem cell factor or a lineage-specifying transcription factor.
Collapse
Affiliation(s)
- Yongqing Liu
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Partha Mukhopadhyay
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - M. Michele Pisano
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Xiaoqin Lu
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Li Huang
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
- School of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Qingxian Lu
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Douglas C. Dean
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Biochemistry and Molecular Biology, University of Louisville Health Sciences Center, Louisville, KY 40202
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| |
Collapse
|
167
|
Roy N, Bommi PV, Bhat UG, Bhattacharjee S, Elangovan I, Li J, Patra KC, Kopanja D, Blunier A, Benya R, Bagchi S, Raychaudhuri P. DDB2 suppresses epithelial-to-mesenchymal transition in colon cancer. Cancer Res 2013; 73:3771-3782. [PMID: 23610444 PMCID: PMC3686976 DOI: 10.1158/0008-5472.can-12-4069] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Colon cancer is one of the deadliest cancers worldwide because of its metastasis to other essential organs. Metastasis of colon cancer involves a complex set of events, including epithelial-to-mesenchymal transition (EMT) that increases invasiveness of the tumor cells. Here, we show that the xeroderma pigmentosum group E (XPE) gene product, damaged DNA-binding protein (DDB)-2, is downregulated in high-grade colon cancers, and it plays a dominant role in the suppression of EMT of the colon cancer cells. Depletion of DDB2 promotes mesenchymal phenotype, whereas expression of DDB2 promotes epithelial phenotype. DDB2 constitutively represses genes that are the key activators of EMT, indicating that DDB2 is a master regulator of EMT of the colon cancer cells. Moreover, we observed evidence that DDB2 functions as a barrier for EMT induced by hypoxia and TGF-β. Also, we provide evidence that DDB2 inhibits metastasis of colon cancer. The results presented here identify a transcriptional regulatory pathway of DDB2 that is directly linked to the mechanisms that suppress metastasis of colon cancer.
Collapse
Affiliation(s)
- Nilotpal Roy
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| | - Prashant V. Bommi
- Center of Molecular Biology of Oral Diseases (M/C 860) College of Dentistry, Cancer Center, University of Illinois at Chicago, 801 S. Paulina Ave, Chicago, IL-60612
| | - Uppoor G. Bhat
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| | - Shaumick Bhattacharjee
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| | - Indira Elangovan
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| | - Jing Li
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| | - Krushna C. Patra
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| | - Dragana Kopanja
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| | - Adam Blunier
- Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, Chicago, Il 60612
| | - Richard Benya
- Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, Chicago, Il 60612
| | - Srilata Bagchi
- Center of Molecular Biology of Oral Diseases (M/C 860) College of Dentistry, Cancer Center, University of Illinois at Chicago, 801 S. Paulina Ave, Chicago, IL-60612
| | - Pradip Raychaudhuri
- Department of Biochemistry and Molecular Genetics (M/C 669), Cancer Center, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL-60607
| |
Collapse
|
168
|
Aldave AJ, Han J, Frausto RF. Genetics of the corneal endothelial dystrophies: an evidence-based review. Clin Genet 2013; 84:109-19. [PMID: 23662738 DOI: 10.1111/cge.12191] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 05/08/2013] [Accepted: 05/08/2013] [Indexed: 11/27/2022]
Abstract
The aim of this review was to provide an evidenced-based review of the genetic basis of the corneal endothelial dystrophies. A review of the English language peer-reviewed literature describing the molecular genetic basis of posterior polymorphous corneal dystrophy (PPCD), congenital hereditary endothelial dystrophy (CHED), Fuchs endothelial corneal dystrophy (FECD) and X-linked endothelial corneal dystrophy (XECD) was performed. Mutations in several genes have been implicated as playing a pathogenic role in the corneal endothelial dystrophies: VSX1 mutations in PPCD1; COL8A2 mutations in PPCD2 and FECD; ZEB1 mutations in PPCD3 and FECD; and SLC4A11 mutations in CHED2 and FECD. However, linkage, association and familial segregation analyses support a role of only one gene in each corneal endothelial dystrophy: ZEB1 in PPCD3, SLC4A11 in CHED2 and COL8A2 in FECD (early onset). In addition, insufficient evidence exists to consider the autosomal dominant form of CHED (CHED1) as distinct from PPCD. An accurate classification of the corneal endothelial dystrophies requires a critical review of the evidence to support the role of each suggested chromosomal locus, gene and genetic mutation associated with a corneal endothelial dystrophy. Only after the separation of evidence from opinion is performed can a critical examination of the molecular pathways that lead to endothelial dysfunction in each of these disorders be accurately performed.
Collapse
Key Words
- corneal dystrophy, Fuchs endothelial, 1
- corneal dystrophy, Fuchs endothelial, 2
- corneal dystrophy, Fuchs endothelial, early onset
- corneal dystrophy, Fuchs endothelial, late onset
- corneal dystrophy, posterior polymorphous, 1
- corneal dystrophy, posterior polymorphous, 2
- corneal dystrophy, posterior polymorphous, 3
- corneal endothelial dystrophy 1
- corneal endothelial dystrophy 2
Collapse
Affiliation(s)
- A J Aldave
- The Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095–7003, USA.
| | | | | |
Collapse
|
169
|
Qiao B, Chen Z, Hu F, Tao Q, Lam AK. BMI-1 activation is crucial in hTERT-induced epithelial-mesenchymal transition of oral epithelial cells. Exp Mol Pathol 2013; 95:57-61. [PMID: 23712029 DOI: 10.1016/j.yexmp.2013.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/17/2013] [Indexed: 11/17/2022]
Abstract
BMI-1 (B lymphoma Mo-MLV insertion region 1 homolog) has been reported to be over-expressed in cell immortalisation and the epithelial-mesenchymal transition (EMT) of cancer cells. The aim of this study is to study the roles of BMI-1 in the human telomerase reverse transcriptase (hTERT)-induced immortalisation and EMT. In this study, hTERT(+)-OME cells and hTERT(+)-HaCaT cells were acquired by viral transduction of hTERT to primary cultured oral keratinocytes and HaCaT cells (skin epidermal cells). siRNA transduction was used for the inhibition of BMI-1 expression. RT-PCR and Western blots were performed to detect the expressions of twist, vimentin, BMI-1, hTERT and p16INK4a in these cell lines. EMT was assessed by immunohistochemistry (expressions of cytokertin & vimentin), Western blots (expressions of Twist, vimentin & E-cadherin) and RT-PCR (expression of Twist). The results indicated that hTERT(+)-OME cells and hTERT(+)-HaCaT cells underwent EMT spontaneously with high expression of Twist. p16INK4a was silenced in both hTERT-transduced cells but could be detected in HaCaT cells. Moreover, BMI-1 was highly expressed in hTERT(+)-OME and hTERT(+)-HaCaT cells but was negative in HaCaT cells. When the expression of BMI-1 was blocked by siRNA transduction, the proliferations of hTERT(+)-OME and hTERT(+)-HaCaT cells were inhibited and the mono-spheroid colony formation of these hTERT-transduced cells was decreased. In addition, the expression of p16INK4a was regained while the expressions of EMT markers (twist and vimentin) were down-regulated in these two BMI-1 blocking cell lines. To conclude, this study suggests BMI-1 expression plays a role in hTERT-induced immortalisation and EMT.
Collapse
Affiliation(s)
- Bin Qiao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | | | | | | | | |
Collapse
|
170
|
Hu Y, He K, Wang D, Yuan X, Liu Y, Ji H, Song J. TMEPAI regulates EMT in lung cancer cells by modulating the ROS and IRS-1 signaling pathways. Carcinogenesis 2013; 34:1764-72. [PMID: 23615405 DOI: 10.1093/carcin/bgt132] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) has been implicated in various pathophysiological processes, including cancer cell migration and distal metastasis. Reactive oxygen species (ROS) and insulin receptor substrate-1 (IRS-1) are important in cancer progression and regulation of EMT. To explore the biological significance and regulatory mechanism of EMT, we determined the expression, the biological function and the signaling pathway of prostate transmembrane protein, androgen induced-1 (TMEPAI), during the induction of EMT and cell migration. Transforming growth factor (TGF)-β1 significantly upregulated the expression of TMEPAI during EMT in human lung adenocarcinoma. Depletion of TMEPAI abolished TGF-β1-induced downregulation of ferritin heavy chain and the subsequent generation of ROS, thus suppressing TGF-β1-induced EMT and cell migration. In addition, increased ROS production and overexpression of TMEPAI downregulated the level of IRS-1. Both the addition of H2O2 and IRS-1 small interfering RNA rescued the ability of TGF-β1 to induce EMT in TMEPAI-depleted cells. Remarkably, the levels of TMEPAI in lung tumor tissues are very high, whereas its expression in normal lung epithelium is very low. Moreover, TMEPAI expression was positively correlated with the cell mesenchymal phenotype and migration potential. Our work reveals that TMEPAI contributes to TGF-β1-induced EMT through ROS production and IRS-1 downregulation in lung cancer cells.
Collapse
Affiliation(s)
- Ying Hu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|
171
|
Wang Y, Zhou BP. Epithelial-mesenchymal Transition---A Hallmark of Breast Cancer Metastasis. ACTA ACUST UNITED AC 2013; 1:38-49. [PMID: 24611128 DOI: 10.1166/ch.2013.1004] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a highly conserved cellular program that converts polarized, immotile epithelial cells to migratory mesenchymal cells. In addition, EMT was initially recognized as a key step for morphogenesis during embryonic development. Emerging evidences indicate that this important developmental program promotes metastasis, drug resistance, and tumor recurrence, features that are associated with a poor clinical outcome for patients with breast cancer. Therefore, better understanding of regulation and signaling pathways in EMT is essential to develop novel targeted therapeutics. In this review, we present updated developments underlying EMT in tumor progression and metastasis, and discuss the challenges remaining in breast cancer research.
Collapse
Affiliation(s)
- Yifan Wang
- Departments of Molecular and Cellular Biochemistry, and Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY, 40506
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and Markey Cancer Center, University of Kentucky School of Medicine, Lexington, KY, 40506
| |
Collapse
|
172
|
Liu Y, Sánchez-Tilló E, Lu X, Huang L, Clem B, Telang S, Jenson AB, Cuatrecasas M, Chesney J, Postigo A, Dean DC. Sequential inductions of the ZEB1 transcription factor caused by mutation of Rb and then Ras proteins are required for tumor initiation and progression. J Biol Chem 2013; 288:11572-80. [PMID: 23443660 DOI: 10.1074/jbc.m112.434951] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Rb1 restricts cell cycle progression, and it imposes cell contact inhibition to suppress tumor outgrowth. It also triggers oncogene-induced senescence to block Ras mutation. Loss of the Rb1 pathway, which is a hallmark of cancer cells, then provides a permissive environment for Ras mutation, and Ras is sufficient for invasive tumor formation in Rb1 family mutant mouse embryo fibroblasts (MEFs). These results demonstrate that sequential mutation of the Rb1 and Ras pathways comprises a tumor initiation axis. Both Rb1 and Ras regulate expression of the transcription factor ZEB1, thereby linking tumor initiation to the subsequent invasion and metastasis, which is induced by ZEB1. ZEB1 acts in a negative feedback loop to block expression of miR-200, which is thought to facilitate tumor invasion and metastasis. However, ZEB1 also represses cyclin-dependent kinase (cdk) inhibitors to control the cell cycle; its mutation in MEFs leads to induction of these inhibitors and premature senescence. Here, we provide evidence for two sequential inductions of ZEB1 during Ras transformation of MEFs. Rb1 constitutively represses cdk inhibitors, and induction of ZEB1 when the Rb1 pathway is lost is required to maintain this repression, allowing for the classic immortalization and loss of cell contact inhibition seen when the Rb1 pathway is lost. In vivo, we show that this induction of ZEB1 is required for Ras-initiated tumor formation. ZEB1 is then further induced by Ras, beyond the level seen with Rb1 mutation, and this Ras superinduction is required to reach a threshold of ZEB1 sufficient for repression of miR-200 and tumor invasion.
Collapse
Affiliation(s)
- Yongqing Liu
- Molecular Targets Program, University of Louisville Health Sciences Center, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Yu W, Zhang Y, Ruest LB, Svoboda KKH. Analysis of Snail1 function and regulation by Twist1 in palatal fusion. Front Physiol 2013; 4:12. [PMID: 23424071 PMCID: PMC3575576 DOI: 10.3389/fphys.2013.00012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 01/10/2013] [Indexed: 12/24/2022] Open
Abstract
Palatal fusion is a tightly controlled process which comprises multiple cellular events, including cell movement and differentiation. Midline epithelial seam (MES) degradation is essential to palatal fusion. In this study, we analyzed the function of Snail1 during the degradation of the MES. We also analyzed the mechanism regulating the expression of the Snail1 gene in palatal shelves. Palatal explants treated with Snail1 siRNA did not degrade the MES and E-cadherin was not repressed leading to failure of palatal fusion. Transforming growth factor beta 3 (Tgfβ3) regulated Snail1 mRNA, as Snail1 expression decreased in response to Tgfβ3 neutralizing antibody and a PI-3 kinase (PI3K) inhibitor. Twist1, in collaboration with E2A factors, regulated the expression of Snail1. Twist1/E47 dimers bond to the Snail1 promoter to activate expression. Without E47, Twist1 repressed Snail1 expression. These results support the hypothesis that Tgfβ3 may signal through Twist1 and then Snail1 to downregulate E-cadherin expression during palatal fusion.
Collapse
Affiliation(s)
- Wenli Yu
- Department of Biomedical Sciences, Center for Craniofacial Research and Diagnosis, Texas A&M University, Baylor College of Dentistry Dallas, TX, USA
| | | | | | | |
Collapse
|
174
|
Voon DCC, Wang H, Koo JKW, Nguyen TAP, Hor YT, Chu YS, Ito K, Fukamachi H, Chan SL, Thiery JP, Ito Y. Runx3 protects gastric epithelial cells against epithelial-mesenchymal transition-induced cellular plasticity and tumorigenicity. Stem Cells 2013; 30:2088-99. [PMID: 22899304 DOI: 10.1002/stem.1183] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The transcription factor RUNX3 functions as a tumor suppressor in the gastrointestinal epithelium, where its loss is an early event in carcinogenesis. While RUNX3 acts concurrently as a mediator of TGF-β signaling and an antagonist of Wnt, the cellular changes that follow its loss and their contribution to tumorigenicity are not fully understood. Here, we report that the loss of Runx3 in gastric epithelial cells results in spontaneous epithelial-mesenchymal transition (EMT). This produces a tumorigenic stem cell-like subpopulation, which remarkably expresses the gastric stem cell marker Lgr5. This phenomenon is due to the compounding effects of the dysregulation of the TGF-β and Wnt pathways. Specifically, Runx3(-/-) p53(-/-) gastric epithelial cells were unexpectedly sensitized for TGF-β-induced EMT, during which the resultant induction of Lgr5 was enhanced by an aberrantly activated Wnt pathway. These data demonstrate a protective role for RUNX3 in safeguarding gastric epithelial cells against aberrant growth factor signaling and the resultant cellular plasticity and stemness.
Collapse
Affiliation(s)
- Dominic Chih-Cheng Voon
- The Cancer Biology Program, Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
ZEB1 imposes a temporary stage-dependent inhibition of muscle gene expression and differentiation via CtBP-mediated transcriptional repression. Mol Cell Biol 2013; 33:1368-82. [PMID: 23339872 DOI: 10.1128/mcb.01259-12] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle development is orchestrated by the myogenic regulatory factor MyoD, whose activity is blocked in myoblasts by proteins preventing its nuclear translocation and/or binding to G/C-centered E-boxes in target genes. Recent evidence indicates that muscle gene expression is also regulated at the cis level by differential affinity for DNA between MyoD and other E-box binding proteins during myogenesis. MyoD binds to G/C-centered E-boxes, enriched in muscle differentiation genes, in myotubes but not in myoblasts. Here, we used cell-based and in vivo Drosophila, Xenopus laevis, and mouse models to show that ZEB1, a G/C-centered E-box binding transcriptional repressor, imposes a temporary stage-dependent inhibition of muscle gene expression and differentiation via CtBP-mediated transcriptional repression. We found that, contrary to MyoD, ZEB1 binds to G/C-centered E-boxes in muscle differentiation genes at the myoblast stage but not in myotubes. Its knockdown results in precocious expression of muscle differentiation genes and acceleration of myotube formation. Inhibition of muscle genes by ZEB1 occurs via transcriptional repression and involves recruitment of the CtBP corepressor. Lastly, we show that the pattern of gene expression associated with muscle differentiation is accelerated in ZEB1(-/-) mouse embryos. These results set ZEB1 as an important regulator of the temporal pattern of gene expression controlling muscle differentiation.
Collapse
|
176
|
Sulforaphane regulates self-renewal of pancreatic cancer stem cells through the modulation of Sonic hedgehog-GLI pathway. Mol Cell Biochem 2012; 373:217-27. [PMID: 23129257 DOI: 10.1007/s11010-012-1493-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 10/17/2012] [Indexed: 12/28/2022]
Abstract
Sulforaphane (SFN), a component of dietary cruciferous vegetables has been characterized for its anti-proliferative properties. We have recently demonstrated that pancreatic CSCs display activation of sonic hedgehog pathway which are fundamental drivers of stem cell renewal, and SFN inhibits the self-renewal of pancreatic CSCs in vitro. Consistent with these observations, we sought to determine the chemopreventive potential of SFN in an in vivo setting. We show here for the first time that sulforaphane treatment resulted in a significant reduction in the tumor growth of orthotopically implanted primary pancreatic CSCs isolated from human pancreatic tumors into the pancreas of NOD/SCID/IL2Rgamma mice, which is mediated through the modulation of Sonic hedgehog-GLI signaling. Hedgehog pathway blockade by SFN at a dose of 20 mg/kg resulted in a 45 % reduction in growth of pancreatic cancer tumors and reduced expression of Shh pathway components, Smo, Gli 1, and Gli 2 in mouse tissues. Further, SFN inhibited the expression of pluripotency maintaining transcription factors Nanog and Oct-4 and angiogenic markers VEGF and PDGFRα which are downstream targets of Gli transcription. Furthermore, SFN treatment resulted in a significant reduction in EMT markers Zeb-1, which correlated with increase in E-Cadherin expression suggesting the blockade of signaling involved in early metastasis. Interestingly, SFN downregulated the expression of Bcl-2 and XIAP to induce apoptosis. These data demonstrate that, at a tolerable dose, inhibition of Shh pathway by SFN results in marked reduction in EMT, metastatic, angiogenic markers with significant inhibition in tumor growth in mice. Since aberrant Shh signaling occurs in pancreatic tumorigenesis, therapeutics that target Shh pathway may improve the outcomes of patients with pancreatic cancer by targeting CSCs, thus suggesting the use of sulforaphane to further improve preventive and therapeutic approaches in patients with this devastating disease.
Collapse
|
177
|
Differential expression of SUMO-specific protease 7 variants regulates epithelial-mesenchymal transition. Proc Natl Acad Sci U S A 2012; 109:17466-71. [PMID: 23045645 DOI: 10.1073/pnas.1209378109] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Two Sentrin/small ubiquitin-like modifier (SUMO)-specific protease 7 (SENP7) variants are naturally expressed in breast epithelia. Breast cancer (BCa) onset down-regulates the short SENP7 splice variant (SENP7S) and enhances the long transcript (SENP7L). Here, we show that SENP7L induction promotes gene expression profiles that favor aberrant proliferation and initiate epithelial-mesenchymal transition (EMT). SENP7L exhibits an interaction domain for the epigenetic remodeler heterochromatin protein 1 α (HP1α) and isopeptidase activity against SUMO-modified HP1α. Loss of this interaction domain, as observed with SENP7S, favors HP1α SUMOylation. SUMOylated HP1α is enriched at E2F-responsive and mesenchymal gene promoters, silences transcription of these genes, and promotes cellular senescence. Elevated SENP7L renders HP1α hypo-SUMOylated, which relieves transcriptional repression of the same genes and concurrently decreases transcription of epithelial-promoting genes via an HP1α-independent mechanism. Consequently, SENP7L levels correlate with EMT, motility, and invasiveness of BCa cells. Stable knockdown of elevated SENP7L levels lessens the dissemination of highly metastatic BCa cells to the lungs from primary implantation sites in in vivo studies. Thus, differential splicing of the SENP7 regulates either tumor suppression or progression.
Collapse
|
178
|
Sánchez-Tilló E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A, Postigo A. EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci 2012; 69:3429-56. [PMID: 22945800 PMCID: PMC11115078 DOI: 10.1007/s00018-012-1122-2] [Citation(s) in RCA: 406] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 12/13/2022]
Abstract
Cancer is a complex multistep process involving genetic and epigenetic changes that eventually result in the activation of oncogenic pathways and/or inactivation of tumor suppressor signals. During cancer progression, cancer cells acquire a number of hallmarks that promote tumor growth and invasion. A crucial mechanism by which carcinoma cells enhance their invasive capacity is the dissolution of intercellular adhesions and the acquisition of a more motile mesenchymal phenotype as part of an epithelial-to-mesenchymal transition (EMT). Although many transcription factors can trigger it, the full molecular reprogramming occurring during an EMT is mainly orchestrated by three major groups of transcription factors: the ZEB, Snail and Twist families. Upregulated expression of these EMT-activating transcription factors (EMT-ATFs) promotes tumor invasiveness in cell lines and xenograft mice models and has been associated with poor clinical prognosis in human cancers. Evidence accumulated in the last few years indicates that EMT-ATFs also regulate an expanding set of cancer cell capabilities beyond tumor invasion. Thus, EMT-ATFs have been shown to cooperate in oncogenic transformation, regulate cancer cell stemness, override safeguard programs against cancer like apoptosis and senescence, determine resistance to chemotherapy and promote tumor angiogenesis. This article reviews the expanding portfolio of functions played by EMT-ATFs in cancer progression.
Collapse
Affiliation(s)
- Ester Sánchez-Tilló
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
| | - Yongqing Liu
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Oriol de Barrios
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Siles
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Lucia Fanlo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- Master Program in Biomedical Research, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Miriam Cuatrecasas
- Department of Pathology, Hospital Clinic and IDIBAPS’ Tumor Bank, 08036 Barcelona, Spain
| | - Douglas S. Darling
- Department of Oral Health and Rehabilitation, Center for Genetics and Molecular Medicine, University of Louisville, Louisville, KY 40202 USA
| | - Douglas C. Dean
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Antoni Castells
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- Institute of Digestive and Metabolic Diseases, Hospital Clinic, 08036 Barcelona, Spain
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- ICREA, 08010 Barcelona, Spain
| |
Collapse
|
179
|
Arnold P, Schöler A, Pachkov M, Balwierz PJ, Jørgensen H, Stadler MB, van Nimwegen E, Schübeler D. Modeling of epigenome dynamics identifies transcription factors that mediate Polycomb targeting. Genome Res 2012; 23:60-73. [PMID: 22964890 PMCID: PMC3530684 DOI: 10.1101/gr.142661.112] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although changes in chromatin are integral to transcriptional reprogramming during cellular differentiation, it is currently unclear how chromatin modifications are targeted to specific loci. To systematically identify transcription factors (TFs) that can direct chromatin changes during cell fate decisions, we model the relationship between genome-wide dynamics of chromatin marks and the local occurrence of computationally predicted TF binding sites. By applying this computational approach to a time course of Polycomb-mediated H3K27me3 marks during neuronal differentiation of murine stem cells, we identify several motifs that likely regulate the dynamics of this chromatin mark. Among these, the sites bound by REST and by the SNAIL family of TFs are predicted to transiently recruit H3K27me3 in neuronal progenitors. We validate these predictions experimentally and show that absence of REST indeed causes loss of H3K27me3 at target promoters in trans, specifically at the neuronal progenitor state. Moreover, using targeted transgenic insertion, we show that promoter fragments containing REST or SNAIL binding sites are sufficient to recruit H3K27me3 in cis, while deletion of these sites results in loss of H3K27me3. These findings illustrate that the occurrence of TF binding sites can determine chromatin dynamics. Local determination of Polycomb activity by REST and SNAIL motifs exemplifies such TF based regulation of chromatin. Furthermore, our results show that key TFs can be identified ab initio through computational modeling of epigenome data sets using a modeling approach that we make readily accessible.
Collapse
Affiliation(s)
- Phil Arnold
- Biozentrum of the University of Basel and Swiss Institute of Bioinformatics, CH 4056 Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
180
|
The T box transcription factor TBX2 promotes epithelial-mesenchymal transition and invasion of normal and malignant breast epithelial cells. PLoS One 2012; 7:e41355. [PMID: 22844464 PMCID: PMC3402503 DOI: 10.1371/journal.pone.0041355] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 06/20/2012] [Indexed: 12/18/2022] Open
Abstract
The T box transcription factor TBX2, a master regulator of organogenesis, is aberrantly amplified in aggressive human epithelial cancers. While it has been shown that overexpression of TBX2 can bypass senescence, a failsafe mechanism against cancer, its potential role in tumor invasion has remained obscure. Here we demonstrate that TBX2 is a strong cell-autonomous inducer of the epithelial-mesenchymal transition (EMT), a latent morphogenetic program that is key to tumor progression from noninvasive to invasive malignant states. Ectopic expression of TBX2 in normal HC11 and MCF10A mammary epithelial cells was sufficient to induce morphological, molecular, and behavioral changes characteristic of EMT. These changes included loss of epithelial adhesion and polarity gene (E-cadherin, ß-catenin, ZO1) expression, and abnormal gain of mesenchymal markers (N-cadherin, Vimentin), as well as increased cell motility and invasion. Conversely, abrogation of endogenous TBX2 overexpression in the malignant human breast carcinoma cell lines MDA-MB-435 and MDA-MB-157 led to a restitution of epithelial characteristics with reciprocal loss of mesenchymal markers. Importantly, TBX2 inhibition abolished tumor cell invasion and the capacity to form lung metastases in a Xenograft mouse model. Meta-analysis of gene expression in over one thousand primary human breast tumors further showed that high TBX2 expression was significantly associated with reduced metastasis-free survival in patients, and with tumor subtypes enriched in EMT gene signatures, consistent with a role of TBX2 in oncogenic EMT. ChIP analysis and cell-based reporter assays further revealed that TBX2 directly represses transcription of E-cadherin, a tumor suppressor gene, whose loss is crucial for malignant tumor progression. Collectively, our results uncover an unanticipated link between TBX2 deregulation in cancer and the acquisition of EMT and invasive features of epithelial tumor cells.
Collapse
|
181
|
Hill L, Browne G, Tulchinsky E. ZEB/miR-200 feedback loop: at the crossroads of signal transduction in cancer. Int J Cancer 2012; 132:745-54. [PMID: 22753312 DOI: 10.1002/ijc.27708] [Citation(s) in RCA: 204] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 06/19/2012] [Indexed: 12/17/2022]
Abstract
Embryonic differentiation programs of epithelial-mesenchymal and mesenchymal-epithelial transition (EMT and MET) represent a mechanistic basis for epithelial cell plasticity implicated in cancer. Transcription factors of the ZEB protein family (ZEB1 and ZEB2) and several microRNA species (predominantly miR-200 family members) form a double negative feedback loop, which controls EMT and MET programs in both development and tumorigenesis. In this article, we review crosstalk between the ZEB/miR-200 axis and several signal transduction pathways activated at different stages of tumor development. The close association of ZEB proteins with these pathways is indirect evidence for the involvement of a ZEB/miR-200 loop in tumor initiation, progression and spread. Additionally, the configuration of signaling pathways involving ZEB/miR-200 loop suggests that ZEB1 and ZEB2 may have different, possibly even opposing, roles in some forms of human cancer.
Collapse
Affiliation(s)
- Louise Hill
- Department of Cancer Studies and Molecular Medicine, University of Leicester, United Kingdom
| | | | | |
Collapse
|
182
|
Das S, Becker BN, Hoffmann FM, Mertz JE. Reversal of transforming growth factor-β induced epithelial-to-mesenchymal transition and the ZEB proteins. FIBROGENESIS & TISSUE REPAIR 2012; 5:S28. [PMID: 23259633 PMCID: PMC3368790 DOI: 10.1186/1755-1536-5-s1-s28] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background The dynamic process of epithelial-to-mesenchymal transition (EMT) is a causal event in kidney fibrosis. This cellular phenotypic transition involves activation of transcriptional responses and remodeling of cellular structures to change cellular function. The molecular mechanisms that directly contribute to the re-establishment of the epithelial phenotype are poorly understood. Results Here, we discuss recent studies from our group and other laboratories identifying signaling pathways leading to the reversal of EMT in fibrotic models. We also present evidence that transcriptional factors such as the ZEB proteins are important regulators for reversal of EMT. Conclusion These studies provide insights into cellular plasticity and possible targets for therapeutic intervention.
Collapse
Affiliation(s)
- Shreyasi Das
- Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Bryan N Becker
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, Wisconsin 53792, USA
| | - F Michael Hoffmann
- Laboratory of Genetics, University of Wisconsin School of Medicine and Public Health, 425-G Henry Mall, Madison, Wisconsin 53706, USA ; McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, 1400 University Ave, Madison, Wisconsin 53706, USA
| | - Janet E Mertz
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, 1400 University Ave, Madison, Wisconsin 53706, USA
| |
Collapse
|
183
|
Qiao B, Gopalan V, Chen Z, Smith RA, Tao Q, Lam AKY. Epithelial-mesenchymal transition and mesenchymal-epithelial transition are essential for the acquisition of stem cell properties in hTERT-immortalised oral epithelial cells. Biol Cell 2012; 104:476-89. [DOI: 10.1111/boc.201100077] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 04/18/2012] [Indexed: 11/30/2022]
|
184
|
EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol 2012; 22:194-207. [DOI: 10.1016/j.semcancer.2012.02.013] [Citation(s) in RCA: 354] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 12/24/2022]
|
185
|
Abstract
Why are many metastases differentiated? Invading and disseminating carcinoma cells can undergo an epithelial-mesenchymal transition (EMT), which is associated with a gain of stem cell-like behaviour. Therefore, EMT has been linked to the cancer stem cell concept. However, it is a matter of debate how subsequent mesenchymal-epithelial transition (MET) fits into the metastatic process and whether a MET is essential. In this Opinion article, I propose two principle types of metastatic progression: phenotypic plasticity involving transient EMT-MET processes and intrinsic genetic alterations keeping cells in an EMT and stemness state. This simplified classification integrates clinically relevant aspects of dormancy, metastatic tropism and therapy resistance, and implies perspectives on treatment strategies against metastasis.
Collapse
Affiliation(s)
- Thomas Brabletz
- Department of General and Visceral Surgery and Comprehensive Cancer Center, University of Freiburg Medical Center, Hugstetter Str. 55, 79106 Freiburg, Germany.
| |
Collapse
|
186
|
Cufí S, Vazquez-Martin A, Oliveras-Ferraros C, Quirantes R, Segura-Carretero A, Micol V, Joven J, Bosch-Barrera J, Del Barco S, Martin-Castillo B, Vellon L, Menendez JA. Metformin lowers the threshold for stress-induced senescence: A role for the microRNA-200 family and miR-205. Cell Cycle 2012; 11:1235-46. [DOI: 10.4161/cc.11.6.19665] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
187
|
The molecular biology of brain metastasis. JOURNAL OF ONCOLOGY 2012; 2012:723541. [PMID: 22481931 PMCID: PMC3317231 DOI: 10.1155/2012/723541] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 11/25/2011] [Indexed: 12/18/2022]
Abstract
Metastasis to the central nervous system (CNS) remains a major cause of morbidity and mortality in patients with systemic cancers. Various crucial interactions between the brain environment and tumor cells take place during the development of the cancer at its new location. The rapid expansion in molecular biology and genetics has advanced our knowledge of the underlying mechanisms involved, from invasion to final colonization of new organ tissues. Understanding the various events occurring at each stage should enable targeted drug delivery and individualized treatments for patients, with better outcomes and fewer side effects. This paper summarizes the principal molecular and genetic mechanisms that underlie the development of brain metastasis (BrM).
Collapse
|
188
|
Swindell WR, Johnston A, Sun L, Xing X, Fisher GJ, Bulyk ML, Elder JT, Gudjonsson JE. Meta-profiles of gene expression during aging: limited similarities between mouse and human and an unexpectedly decreased inflammatory signature. PLoS One 2012; 7:e33204. [PMID: 22413003 PMCID: PMC3296693 DOI: 10.1371/journal.pone.0033204] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 02/13/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Skin aging is associated with intrinsic processes that compromise the structure of the extracellular matrix while promoting loss of functional and regenerative capacity. These processes are accompanied by a large-scale shift in gene expression, but underlying mechanisms are not understood and conservation of these mechanisms between humans and mice is uncertain. RESULTS We used genome-wide expression profiling to investigate the aging skin transcriptome. In humans, age-related shifts in gene expression were sex-specific. In females, aging increased expression of transcripts associated with T-cells, B-cells and dendritic cells, and decreased expression of genes in regions with elevated Zeb1, AP-2 and YY1 motif density. In males, however, these effects were contrasting or absent. When age-associated gene expression patterns in human skin were compared to those in tail skin from CB6F1 mice, overall human-mouse correspondence was weak. Moreover, inflammatory gene expression patterns were not induced with aging of mouse tail skin, and well-known aging biomarkers were in fact decreased (e.g., Clec7a, Lyz1 and Lyz2). These unexpected patterns and weak human-mouse correspondence may be due to decreased abundance of antigen presenting cells in mouse tail skin with age. CONCLUSIONS Aging is generally associated with a pro-inflammatory state, but we have identified an exception to this pattern with aging of CB6F1 mouse tail skin. Aging therefore does not uniformly heighten inflammatory status across all mouse tissues. Furthermore, we identified both intercellular and intracellular mechanisms of transcriptome aging, including those that are sex- and species-specific.
Collapse
Affiliation(s)
- William R Swindell
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
189
|
The Roles of Mitogen-Activated Protein Kinase Pathways in TGF-β-Induced Epithelial-Mesenchymal Transition. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:289243. [PMID: 22363839 PMCID: PMC3272823 DOI: 10.1155/2012/289243] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 10/22/2011] [Accepted: 10/23/2011] [Indexed: 02/07/2023]
Abstract
The mitogen-activated protein kinase (MAPK) pathway allows cells to interpret external signals and respond appropriately, especially during the epithelial-mesenchymal transition (EMT). EMT is an important process during embryonic development, fibrosis, and tumor progression in which epithelial cells acquire mesenchymal, fibroblast-like properties and show reduced intercellular adhesion and increased motility. TGF-β signaling is the first pathway to be described as an inducer of EMT, and its relationship with the Smad family is already well characterized. Studies of four members of the MAPK family in different biological systems have shown that the MAPK and TGF-β signaling pathways interact with each other and have a synergistic effect on the secretion of additional growth factors and cytokines that in turn promote EMT. In this paper, we present background on the regulation and function of MAPKs and their cascades, highlight the mechanisms of MAPK crosstalk with TGF-β signaling, and discuss the roles of MAPKs in EMT.
Collapse
|
190
|
Association of TCF4 and CLU polymorphisms with Fuchs' endothelial dystrophy and implication of CLU and TGFBI proteins in the disease process. Eur J Hum Genet 2012; 20:632-8. [PMID: 22234156 DOI: 10.1038/ejhg.2011.248] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fuchs' endothelial dystrophy (FED) is a disease affecting the corneal endothelium. Recent studies reported significant association of polymorphisms in the TCF4 (transcription factor 4) gene, and a borderline association of PTPRG (protein tyrosine phosphatase, receptor type, G) variants with late-onset FED in Caucasians from the United States. Association of TCF4 has also been reported in the Chinese population. We aimed to determine association of the reported polymorphisms in TCF4 and PTPRG, and association of polymorphisms in the candidate genes ZEB1 (zinc-finger E-box binding homoebox 1), COL8A2 (collagen, type VIII, alpha 2), TGFBI (transforming growth factor, β-induced) and CLU (clusterin) in Australian cases. We also compared the expression of TGFBI and CLU proteins between FED and normal whole corneas. In all, 30 single-nucleotide polymorphisms (SNPs) from the candidate genes were genotyped in 103 cases and 275 controls. Each SNP and haplotype was assessed for association with the disease. SNP analysis identified an association of TCF4 (rs613872 (P=5.25 × 10(-15), OR=4.05), rs9954153 (P=3.37 × 10(-7), OR=2.58), rs2286812 (P=4.23 × 10(-6), OR=2.55) and rs17595731 (P=3.57 × 10(-5), OR=3.79)), CLU (rs17466684; P=0.003, OR=1.85) and one haplotype of TGFBI SNPs (P=0.011, OR=2.29) with FED in Caucasian Australians. No evidence for genetic association of PTPRG, ZEB1 and COL8A2 was found. Immunohistochemistry showed differential expression of CLU and TGFBI proteins in FED-affected compared with normal corneas. In conclusion, variation in TCF4, CLU and TGFBI, but not PTPRG, ZEB1 and COL8A2 genes are associated with FED in Caucasian Australian cases. Differential expression of CLU and TGFBI proteins in FED-affected corneas provides novel insights into the disease mechanism.
Collapse
|
191
|
Kobayashi A, Okuda H, Xing F, Pandey PR, Watabe M, Hirota S, Pai SK, Liu W, Fukuda K, Chambers C, Wilber A, Watabe K. Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. J Exp Med 2011; 208:2641-55. [PMID: 22124112 PMCID: PMC3244043 DOI: 10.1084/jem.20110840] [Citation(s) in RCA: 315] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 11/01/2011] [Indexed: 12/20/2022] Open
Abstract
Metastatic disease is the major cause of cancer deaths, and recurrent tumors at distant organs are a critical issue. However, how metastatic tumor cells become dormant and how and why tumors recur in target organs are not well understood. In this study, we demonstrate that BMP7 (bone morphogenetic protein 7) secreted from bone stromal cells induces senescence in prostate cancer stem-like cells (CSCs) by activating p38 mitogen-activated protein kinase and increasing expression of the cell cycle inhibitor, p21, and the metastasis suppressor gene, NDRG1 (N-myc downstream-regulated gene 1). This effect of BMP7 depended on BMPR2 (BMP receptor 2), and BMPR2 expression inversely correlated with recurrence and bone metastasis in prostate cancer patients. Importantly, this BMP7-induced senescence in CSCs was reversible upon withdrawal of BMP7. Furthermore, treatment of mice with BMP7 significantly suppressed the growth of CSCs in bone, whereas the withdrawal of BMP7 restarted growth of these cells. These results suggest that the BMP7-BMPR2-p38-NDRG1 axis plays a critical role in dormancy and recurrence of prostate CSCs in bone and suggest a potential therapeutic utility of BMP7 for recurrent metastatic disease.
Collapse
Affiliation(s)
- Aya Kobayashi
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Hiroshi Okuda
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Fei Xing
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Puspa R. Pandey
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Misako Watabe
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Shigeru Hirota
- Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Sudha K. Pai
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Wen Liu
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Koji Fukuda
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Christopher Chambers
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| | - Kounosuke Watabe
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 19626
| |
Collapse
|
192
|
Li XY, Zhou X, Rowe RG, Hu Y, Schlaepfer DD, Ilić D, Dressler G, Park A, Guan JL, Weiss SJ. Snail1 controls epithelial-mesenchymal lineage commitment in focal adhesion kinase-null embryonic cells. ACTA ACUST UNITED AC 2011; 195:729-38. [PMID: 22105351 PMCID: PMC3257570 DOI: 10.1083/jcb.201105103] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mouse embryonic cells isolated from focal adhesion kinase (FAK)-null animals at embryonic day 7.5 display multiple defects in focal adhesion remodeling, microtubule dynamics, mechanotransduction, proliferation, directional motility, and invasion. To date, the ability of FAK to modulate cell function has been ascribed largely to its control of posttranscriptional signaling cascades in this embryonic cell population. In this paper, we demonstrate that FAK unexpectedly exerts control over an epithelial-mesenchymal transition (EMT) program that commits embryonic FAK-null cells to an epithelial status highlighted by the expression of E-cadherin, desmoplakin, and cytokeratins. FAK rescue reestablished the mesenchymal characteristics of FAK-null embryonic cells to generate committed mouse embryonic fibroblasts via an extracellular signal-related kinase- and Akt-dependent signaling cascade that triggered Snail1 gene expression and Snail1 protein stabilization. These findings indentify FAK as a novel regulator of Snail1-dependent EMT in embryonic cells and suggest that multiple defects in FAK(-/-) cell behavior can be attributed to an inappropriate commitment of these cells to an epithelial, rather than fibroblastic, phenotype.
Collapse
Affiliation(s)
- Xiao-Yan Li
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Anders L, Ke N, Hydbring P, Choi YJ, Widlund HR, Chick JM, Zhai H, Vidal M, Gygi SP, Braun P, Sicinski P. A systematic screen for CDK4/6 substrates links FOXM1 phosphorylation to senescence suppression in cancer cells. Cancer Cell 2011; 20:620-34. [PMID: 22094256 PMCID: PMC3237683 DOI: 10.1016/j.ccr.2011.10.001] [Citation(s) in RCA: 435] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 04/21/2011] [Accepted: 10/02/2011] [Indexed: 12/31/2022]
Abstract
Cyclin D-dependent kinases (CDK4 and CDK6) are positive regulators of cell cycle entry and they are overactive in the majority of human cancers. However, it is currently not completely understood by which cellular mechanisms CDK4/6 promote tumorigenesis, largely due to the limited number of identified substrates. Here we performed a systematic screen for substrates of cyclin D1-CDK4 and cyclin D3-CDK6. We identified the Forkhead Box M1 (FOXM1) transcription factor as a common critical phosphorylation target. CDK4/6 stabilize and activate FOXM1, thereby maintain expression of G1/S phase genes, suppress the levels of reactive oxygen species (ROS), and protect cancer cells from senescence. Melanoma cells, unlike melanocytes, are highly reliant on CDK4/6-mediated senescence suppression, which makes them particularly susceptible to CDK4/6 inhibition.
Collapse
Affiliation(s)
- Lars Anders
- Department of Cancer Biology Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| | - Nan Ke
- Department of Cancer Biology Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
| | - Per Hydbring
- Department of Cancer Biology Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
| | - Yoon J. Choi
- Department of Cancer Biology Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
| | - Hans R. Widlund
- Department of Dermatology Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Joel M. Chick
- Department of Cell Biology Harvard Medical School, Boston, MA 02115, USA
| | - Huili Zhai
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Marc Vidal
- Department of Cancer Biology Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Center for Cancer Systems Biology (CCSB) Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
| | - Stephen P. Gygi
- Department of Cell Biology Harvard Medical School, Boston, MA 02115, USA
| | - Pascal Braun
- Department of Cancer Biology Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Center for Cancer Systems Biology (CCSB) Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
| | - Piotr Sicinski
- Department of Cancer Biology Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Genetics Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
194
|
Xue Y, Li L, Zhang D, Wu K, Chen Y, Zeng J, Wang X, He D. Twisted epithelial-to-mesenchymal transition promotes progression of surviving bladder cancer T24 cells with hTERT-dysfunction. PLoS One 2011; 6:e27748. [PMID: 22110753 PMCID: PMC3216997 DOI: 10.1371/journal.pone.0027748] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 10/24/2011] [Indexed: 12/21/2022] Open
Abstract
Background Human cancer cells maintain telomeres to protect cells from senescence through telomerase activity (TA) or alternative lengthening of telomeres (ALT) in different cell types. Moreover, cellular senescence can be bypassed by Epithelial-to-mesenchymal transition (EMT) during cancer progression in diverse solid tumors. However, it has not been elucidated the characteristics of telomere maintenance and progression ability after long-term culture in bladder cancer T24 cells with hTERT dysfunction. Methodology/Principal Findings In this study, by using a dominant negative mutant human telomerase reverse transcriptase (hTERT) vector to inhibit TA in bladder cancer T24 cells, we observed the appearance of long phenotype of telomere length and the ALT-associated PML body (APB) complex after the 27th passage, indicating the occurrence of ALT-like pathway in surviving T24/DN868A cells with telomerase inhibition. Meanwhile, telomerase inhibition resulted in significant EMT as shown by change in cellular morphology concomitant with variation of EMT markers. Consistently, the surviving T24/DN868A cells showed increased progression ability in vitro and in vivo. In addition, we found Twist was activated to mediate EMT in surviving T24/DN868A samples. Conclusions/Significance Taken together, our findings indicate that bladder cancer T24 cells may undergo the telomerase-to-ALT-like conversion and promote cancer progression at advanced stages through promoting EMT, thus providing novel possible insight into the mechanism of resistance to telomerase inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Yan Xue
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lei Li
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dong Zhang
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Kaijie Wu
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yule Chen
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jin Zeng
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xinyang Wang
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dalin He
- Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- * E-mail:
| |
Collapse
|
195
|
Ansieau S, Courtois-Cox S, Morel AP, Puisieux A. Failsafe program escape and EMT: a deleterious partnership. Semin Cancer Biol 2011; 21:392-6. [PMID: 21986518 DOI: 10.1016/j.semcancer.2011.09.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 09/22/2011] [Accepted: 09/30/2011] [Indexed: 12/16/2022]
Abstract
The epithelial to mesenchymal transition (EMT) is a latent embryonic process which can be aberrantly reactivated during tumor progression. It is generally viewed as one of the main forces driving metastatic dissemination, by providing cells with invasive and motility capabilities. The aberrant reactivation of embryonic EMT inducers has now been additionally linked to escape from senescence and apoptosis, which suggests a role in tumor initiation. This oncogenic potential relies on the ability of EMT inducers to neutralize both the RB and p53 oncosuppressive pathways. RB and p53 have recently been described as key factors in the maintenance of epithelial morphology, which suggests an unexpected and intimate crosstalk between EMT and the corresponding safety programs. In this review, we attempt to understand how these two cell processes are interlinked and might facilitate cell transformation and tumor initiation.
Collapse
Affiliation(s)
- Stéphane Ansieau
- Inserm UMR-S1052, Centre de Recherche en Cancérologie, Lyon F-69008, France.
| | | | | | | |
Collapse
|
196
|
Ohashi S, Natsuizaka M, Naganuma S, Kagawa S, Kimura S, Itoh H, Kalman RA, Nakagawa M, Darling DS, Basu D, Gimotty PA, Klein-Szanto AJ, Diehl JA, Herlyn M, Nakagawa H. A NOTCH3-mediated squamous cell differentiation program limits expansion of EMT-competent cells that express the ZEB transcription factors. Cancer Res 2011; 71:6836-47. [PMID: 21890822 DOI: 10.1158/0008-5472.can-11-0846] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Zinc finger E-box-binding (ZEB) proteins ZEB1 and ZEB2 are transcription factors essential in TGF-β-mediated senescence, epithelial-to-mesenchymal transition (EMT), and cancer stem cell functions. ZEBs are negatively regulated by members of the miR-200 microRNA family, but precisely how tumor cells expressing ZEBs emerge during invasive growth remains unknown. Here, we report that NOTCH3-mediated signaling prevents expansion of a unique subset of ZEB-expressing cells. ZEB expression was associated with the lack of cellular capability of undergoing NOTCH3-mediated squamous differentiation in human esophageal cells. Genetic inhibition of the Notch-mediated transcriptional activity by dominant-negative Mastermind-like 1 (DNMAML1) prevented squamous differentiation and induction of Notch target genes including NOTCH3. Moreover, DNMAML1-enriched EMT-competent cells exhibited robust upregulation of ZEBs, downregulation of the miR-200 family, and enhanced anchorage-independent growth and tumor formation in nude mice. RNA interference experiments suggested the involvement of ZEBs in anchorage-independent colony formation, invasion, and TGF-β-mediated EMT. Invasive growth and impaired squamous differentiation were recapitulated upon Notch inhibition by DNMAML1 in organotypic three-dimensional culture, a form of human tissue engineering. Together, our findings indicate that NOTCH3 is a key factor limiting the expansion of ZEB-expressing cells, providing novel mechanistic insights into the role of Notch signaling in the cell fate regulation and disease progression of esophageal squamous cancers.
Collapse
Affiliation(s)
- Shinya Ohashi
- Gastroenterology Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Sánchez-Tilló E, Siles L, de Barrios O, Cuatrecasas M, Vaquero EC, Castells A, Postigo A. Expanding roles of ZEB factors in tumorigenesis and tumor progression. Am J Cancer Res 2011; 1:897-912. [PMID: 22016835 PMCID: PMC3196287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 08/19/2011] [Indexed: 05/31/2023] Open
Abstract
The ZEB family of transcription factors regulates key factors during embryonic development and cell differentiation but their role in cancer biology has only more recently begun to be recognized. Early evidence showed that ZEB proteins induce an epithelial-to-mesenchymal transition linking their expression with increased aggressiveness and metastasis in mice models and a wide range of primary human carcinomas. Reports over the last few years have found that ZEB proteins also play critical roles in the maintenance of cancer cell stemness, control of replicative senescence, tumor angiogenesis, overcoming of oncogenic addiction and resistance to chemotherapy. These expanding roles in tumorigenesis and tumor progression set ZEB proteins as potential diagnostic, prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Ester Sánchez-Tilló
- Group of Transcriptional Regulation of Gene Expression, Dept. of Oncology and Hematology, IDIBAPSBarcelona, Spain
| | - Laura Siles
- Master Program in Molecular Biotechnology, University of BarcelonaSpain
| | | | | | - Eva C Vaquero
- Dept. of Gastroenterology, Hospital Clinic of Barcelona, CIBERehd, IDIBAPSBarcelona, Spain
| | - Antoni Castells
- Dept. of Gastroenterology, Hospital Clinic of Barcelona, CIBERehd, IDIBAPSBarcelona, Spain
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Dept. of Oncology and Hematology, IDIBAPSBarcelona, Spain
- ICREABarcelona, Spain
- James Graham Brown Cancer Center, University of LouisvilleKY, USA
| |
Collapse
|
198
|
Nieto MA. The ins and outs of the epithelial to mesenchymal transition in health and disease. Annu Rev Cell Dev Biol 2011; 27:347-76. [PMID: 21740232 DOI: 10.1146/annurev-cellbio-092910-154036] [Citation(s) in RCA: 567] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The epithelial to mesenchymal transition (EMT) converts epithelial cells into migratory and invasive cells and is a fundamental event in morphogenesis. Although its relevance in the progression of cancer and organ fibrosis had been debated until recently, the EMT is now established as an important step in the metastatic cascade of epithelial tumors. The similarities between pathological and developmental EMTs validate the embryo as the best model to understand the molecular and cellular mechanisms involved in this process, identifying those that are hijacked during the progression of cancer and organ degeneration. Our ever-increasing understanding of how transcription factors regulate the EMT has revealed complex regulatory loops coupled to posttranscriptional and epigenetic regulatory programs. The EMT is now integrated into the systemic activities of whole organisms, establishing links with cell survival, stemness, inflammation, and immunity. In addition, the EMT now constitutes a promising target for the treatment of cancer and organ-degenerative diseases.
Collapse
Affiliation(s)
- M Angela Nieto
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, San Juan de Alicante 03550, Spain.
| |
Collapse
|
199
|
Vazquez-Martin A, López-Bonetc E, Cufí S, Oliveras-Ferraros C, Del Barco S, Martin-Castillo B, Menendez JA. Repositioning chloroquine and metformin to eliminate cancer stem cell traits in pre-malignant lesions. Drug Resist Updat 2011; 14:212-23. [PMID: 21600837 DOI: 10.1016/j.drup.2011.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 01/07/2023]
Abstract
Ideal oncology drugs would be curative after a short treatment course if they could eliminate epithelium-originated carcinomas at their non-invasive, pre-malignant stages. Such ideal molecules, which are expected to molecularly abrogate all the instrumental mechanisms acquired by migrating cancer stem cells (CSCs) to by-pass tumour suppressor barriers, might already exist. We here illustrate how system biology strategies for repositioning existing FDA-approved drugs may accelerate our therapeutic capacity to eliminate CSC traits in pre-invasive intraepithelial neoplasias. First, we describe a signalling network signature that overrides bioenergetics stress- and oncogene-induced senescence (OIS) phenomena in CSCs residing at pre-invasive lesions. Second, we functionally map the anti-malarial chloroquine and the anti-diabetic metformin ("old drugs") to their recently recognized CSC targets ("new uses") within the network. By discussing the preclinical efficacy of chloroquine and metformin to inhibiting the genesis and self-renewal of CSCs we finally underscore the expected translational impact of the "old drugs-new uses" repurposing strategy to open a new CSC-targeted chemoprevention era.
Collapse
Affiliation(s)
- Alejandro Vazquez-Martin
- Unit of Translational Research, Catalan Institute of Oncology-Girona, Avenida de Francia s/n, E-17007 Girona, Catalonia, Spain
| | | | | | | | | | | | | |
Collapse
|
200
|
Matuszak EA, Kyprianou N. Androgen regulation of epithelial-mesenchymal transition in prostate tumorigenesis. Expert Rev Endocrinol Metab 2011; 6:469-482. [PMID: 23667383 PMCID: PMC3648215 DOI: 10.1586/eem.11.32] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Prostate cancer patient mortality is ascribed to the spread of cancerous cells to areas outside the prostate gland and the inability of current treatment strategies to effectively block progression to metastasis. Understanding the cellular mechanisms contributing to the dissemination of malignant cells and metastasis is critically significant to the generation of effective therapeutic modalities for improved patient survival while combating therapeutic resistance. In recent years, the phenomenon of epithelial-mesenchymal transitions (EMTs) has received considerable attention due to accumulating evidence indicating a role for this developmentally conserved process in tumorigenesis. Cancer cells at the invasive edges of tumors undergo EMT under the influence of contextual signals that they receive from the microenvironment, such as TGF-β. Also derived from developmental studies is the fact that EMT induction is reversible; thus, upon removal of EMT-inducing signals, cells occasionally revert to the epithelial state of their cellular ancestors via the process of mesenchymal-epithelial transition. This article discusses the current evidence supporting a central role for EMT and its reverse process, mesenchymal-epithelial transition, in the metastatic progression of prostate cancer to advanced disease and the involvement of androgen signaling in its regulation towards the development of castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Emily A Matuszak
- Department of Toxicology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
- Department of Surgery/Urology, University of Kentucky College of Medicine, KY, USA
| | - Natasha Kyprianou
- Department of Toxicology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
- Department of Surgery/Urology, University of Kentucky College of Medicine, KY, USA
- Department of Pathology, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|